Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-37732224

RESUMEN

Tissue resident myeloid cells (TRM) in adults have highly variable lifespans and may be derived from early embryonic yolk sac, fetal liver or bone marrow. Some of these TRM are known pathogenic participants in congenital and acquired diseases. Myeloablative conditioning and hematopoietic stem cell transplant can replace long-lived brain TRM resulting in clinical improvements in metabolic storage diseases. With the advent of antibody-drug-conjugate (ADC) targeted cell killing as a cell selective means of transplant conditioning, we assessed the impact of anti-CD45-ADC on TRM in multiple tissues. Replacement of TRM ranged from 40 to 95 percent efficiencies in liver, lung, and skin tissues, after a single anti-CD45-ADC dose and bone marrow hematopoietic cell transfer. Of note, the population size of TRM in tissues returned to pre-treatment levels suggesting a regulated control of TRM abundance. As expected, brain, microglia were not affected, but brain monocytes and macrophages were 50% replaced. Anti-CD45-ADC and adoptive cell transfer were then tested in the chronic acquired condition, atherosclerosis exacerbated by Tet2 mutant clonal hematopoiesis. Plaque resident myeloid cells were efficiently replaced with anti-CD45-ADC and wild-type bone marrow cells. Notably, this reduced existent atherosclerotic plaque burden. Overall, these results indicate that anti-CD45-ADC clears both HSC and TRM niches enabling cell replacement to achieve disease modification in a resident myeloid cell driven disease.

2.
Blood Adv ; 7(22): 6964-6973, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37748049

RESUMEN

Tissue-resident myeloid (TRM) cells in adults have highly variable lifespans, and may be derived from early embryonic yolk sac, fetal liver, or bone marrow. Some of these TRM cells are known pathogenic participants in congenital and acquired diseases. Myeloablative conditioning and hematopoietic stem cell transplantation can replace long-lived brain TRM cells, resulting in clinical improvements in metabolic storage diseases. With the advent of antibody-drug conjugate (ADC)-targeted cell killing as a cell-selective means of transplant conditioning, we assessed the impact of anti-CD45-ADC on TRM cells in multiple tissues. Replacement of TRM cells ranged from 40% to 95% efficiencies in liver, lung, and skin tissues, after a single anti-CD45-ADC dose and bone marrow hematopoietic cell transfer. Of note, the population size of TRM cells in tissues returned to pretreatment levels, suggesting a regulated control of TRM cell abundance. As expected, brain microglia were not affected, but brain monocytes and macrophages were 50% replaced. Anti-CD45-ADC and adoptive cell transfer were then tested in the chronic acquired condition, atherosclerosis exacerbated by Tet2 mutant clonal hematopoiesis. Plaque-resident myeloid cells were efficiently replaced with anti-CD45-ADC and wild-type bone marrow cells. Notably, this reduced existent atherosclerotic plaque burden. Overall, these results indicate that the anti-CD45-ADC clears both hematopoietic stem and TRM cells from their niches, enabling cell replacement to achieve disease modification in a resident myeloid cell-driven disease.


Asunto(s)
Inmunoconjugados , Adulto , Humanos , Inmunoconjugados/farmacología , Macrófagos , Monocitos , Médula Ósea , Microglía
3.
Mol Ther Methods Clin Dev ; 17: 455-464, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32226796

RESUMEN

Conditioning chemotherapy is used to deplete hematopoietic stem cells in the recipient's marrow, facilitating donor cell engraftment. Although effective, a major issue with chemotherapy is the systemic genotoxicity that increases the risk for secondary malignancies. Antibody conjugates targeting hematopoietic cells are an emerging non-genotoxic method of opening the marrow niche and promoting engraftment of transplanted cells while maintaining intact marrow cellularity. Specifically, this platform would be useful in diseases associated with DNA damage or cancer predisposition, such as dyskeratosis congenita, Schwachman-Diamond syndrome, and Fanconi anemia (FA). Our approach utilizes antibody-drug conjugates (ADC) as an alternative conditioning regimen in an FA mouse model of autologous transplantation. Antibodies targeting either CD45 or CD117 were conjugated to saporin (SAP), a ribosomal toxin. FANCA knockout mice were conditioned with either CD45-SAP or CD117-SAP prior to receiving whole marrow from a heterozygous healthy donor. Bone marrow and peripheral blood analysis revealed equivalent levels of donor engraftment, with minimal toxicity in ADC-treated groups as compared with cyclophosphamide-treated controls. Our findings suggest ADCs may be an effective conditioning strategy in stem cell transplantation not only for diseases where traditional chemotherapy is not tolerated, but also more broadly for the field of blood and marrow transplantation.

4.
Nat Methods ; 15(11): 941-946, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30297964

RESUMEN

CRISPR-Cas9 screening allows genome-wide interrogation of gene function. Currently, to achieve the high and uniform Cas9 expression desirable for screening, one needs to engineer stable and clonal Cas9-expressing cells-an approach that is not applicable in human primary cells. Guide Swap permits genome-scale pooled CRISPR-Cas9 screening in human primary cells by exploiting the unexpected finding that editing by lentivirally delivered, targeted guide RNAs (gRNAs) occurs efficiently when Cas9 is introduced in complex with nontargeting gRNA. We validated Guide Swap in depletion and enrichment screens in CD4+ T cells. Next, we implemented Guide Swap in a model of ex vivo hematopoiesis, and identified known and previously unknown regulators of CD34+ hematopoietic stem and progenitor cell (HSPC) expansion. We anticipate that this platform will be broadly applicable to other challenging cell types, and thus will enable discovery in previously inaccessible but biologically relevant human primary cell systems.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , Edición Génica , Genoma Humano , Células Madre Hematopoyéticas/metabolismo , ARN Guía de Kinetoplastida/genética , Linfocitos T CD8-positivos/citología , Células Cultivadas , Células HEK293 , Células Madre Hematopoyéticas/citología , Humanos
5.
SLAS Discov ; 23(7): 697-707, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29843542

RESUMEN

The goal of high-throughput screening is to enable screening of compound libraries in an automated manner to identify quality starting points for optimization. This often involves screening a large diversity of compounds in an assay that preserves a connection to the disease pathology. Phenotypic screening is a powerful tool for drug identification, in that assays can be run without prior understanding of the target and with primary cells that closely mimic the therapeutic setting. Advanced automation and high-content imaging have enabled many complex assays, but these are still relatively slow and low throughput. To address this limitation, we have developed an automated workflow that is dedicated to processing complex phenotypic assays for flow cytometry. The system can achieve a throughput of 50,000 wells per day, resulting in a fully automated platform that enables robust phenotypic drug discovery. Over the past 5 years, this screening system has been used for a variety of drug discovery programs, across many disease areas, with many molecules advancing quickly into preclinical development and into the clinic. This report will highlight a diversity of approaches that automated flow cytometry has enabled for phenotypic drug discovery.


Asunto(s)
Descubrimiento de Drogas , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Automatización de Laboratorios , Plaquetas/efectos de los fármacos , Línea Celular , Biología Computacional/métodos , Análisis de Datos , Descubrimiento de Drogas/instrumentación , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Ensayos Analíticos de Alto Rendimiento/instrumentación , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Hibridomas , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
6.
Cell Stem Cell ; 18(1): 144-55, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26669897

RESUMEN

Clinical application of umbilical cord blood (UCB) as a source of hematopoietic stem cells for transplantation is limited by low CD34+ cell dose, increased risk of graft failure, and slow hematopoietic recovery. While the cell dose limitation is partially mitigated by using two UCB units, larger-dosed single units would be preferable. We have evaluated the feasibility and safety of StemRegenin-1 (SR-1), an aryl hydrocarbon receptor antagonist that expands CD34+ cells, by placing one of the two units in expansion culture. SR-1 produced a 330-fold increase in CD34+ cells and led to engraftment in 17/17 patients at a median of 15 days for neutrophils and 49 days for platelets, significantly faster than in patients treated with unmanipulated UCB. Taken together, the marked expansion, absence of graft failure, and enhanced hematopoietic recovery support testing of SR-1 expansion as a stand-alone graft and suggest it may ameliorate a limitation of UCB transplant.


Asunto(s)
Sangre Fetal/citología , Neoplasias Hematológicas/terapia , Células Madre Hematopoyéticas/citología , Purinas/química , Adolescente , Adulto , Antígenos CD34/metabolismo , Plaquetas/citología , Células Cultivadas , Niño , Trasplante de Células Madre de Sangre del Cordón Umbilical , Criopreservación , Supervivencia de Injerto , Antígenos HLA/metabolismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Persona de Mediana Edad , Neutrófilos/citología , Linfocitos T/citología , Telómero/ultraestructura , Acondicionamiento Pretrasplante , Adulto Joven
7.
Nat Rev Clin Oncol ; 12(3): 163-74, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25511187

RESUMEN

The use of umbilical cord blood (UCB) as an alternative haematopoietic cell source in lieu of bone marrow for haematopoietic reconstitution is increasingly becoming a mainstay treatment for both malignant and nonmalignant diseases, as most individuals will have at least one available, suitably HLA-matched unit of blood. The principal limitation of UCB is the low and finite number of haematopoietic stem and progenitor cells (HSPC) relative to the number found in a typical bone marrow or mobilized peripheral blood allograft, which leads to prolonged engraftment times. In an attempt to overcome this obstacle, strategies that are often based on native processes occurring in the bone marrow microenvironment or 'niche' have been developed with the goal of accelerating UCB engraftment. In broad terms, the two main approaches have been either to expand UCB HSPC ex vivo before transplantation, or to modulate HSPC functionality to increase the efficiency of HSPC homing to the bone marrow niche after transplant both of which enhance the biological activities of the engrafted HSPC. Several early phase clinical trials of these approaches have reported promising results.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal/citología , Células Madre Hematopoyéticas , Investigación Biomédica Traslacional , Técnicas de Cultivo de Célula , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Supervivencia de Injerto , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Pronóstico , Trasplante Homólogo , Resultado del Tratamiento
8.
ACS Chem Biol ; 8(5): 866-70, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23441826

RESUMEN

Hematopoietic stem cells (HSCs) are the progenitor cells that give rise to all blood cells. The ability to control HSC differentiation has the potential to improve the success of bone marrow transplants and the production of functional blood cells ex vivo. Here we performed an unbiased screen using primary human CD34(+) hematopoietic stem and progenitor cells (HSPCs) to identify natural products that selectively control their differentiation. We identified a plant-derived natural product, eupalinilide E, that promotes the ex vivo expansion of HSPCs and hinders the in vitro development of erythrocytes. This activity was additive with aryl hydrocarbon receptor (AhR) antagonists, which are also known to expand HSCs and currently in clinical development. These findings reveal a new activity for eupalinilide E, and suggest that it may be a useful tool to probe the mechanisms of hematopoiesis and improve the ex vivo production of progenitors for therapeutic purposes.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Sesquiterpenos/farmacología , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias/métodos , Medios de Cultivo/química , Medios de Cultivo/farmacología , Dioxinas/farmacología , Evaluación Preclínica de Medicamentos/métodos , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , FN-kappa B/metabolismo , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Transferrina/metabolismo , Sesquiterpenos/química
9.
Proc Natl Acad Sci U S A ; 109(35): 14019-23, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22891346

RESUMEN

Molecules that control the lineage commitment of hematopoietic stem cells (HSCs) may allow the expansion of enriched progenitor populations for both research and therapeutic uses. In an effort to better understand and control the differentiation of HSCs to megakaryocytes, we carried out an image-based screen of a library of 50,000 heterocycles using primary human CD34(+) cells. A class of naphthyridinone derivatives was identified that induces the differentiation of common myeloid progenitors (CMP) to megakaryocytes. Kinase profiling and subsequent functional assays revealed that these compounds act through inhibition of platelet-derived growth factor receptor (PDGFR) signaling in CMPs. Such molecules may ultimately have clinical utility in the treatment of thrombocytopenia.


Asunto(s)
Células Madre Hematopoyéticas/citología , Ensayos Analíticos de Alto Rendimiento/métodos , Megacariocitos/citología , Naftiridinas/farmacología , Trombopoyesis/efectos de los fármacos , Trombopoyesis/fisiología , Antígenos CD34/metabolismo , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Megacariocitos/metabolismo , Microscopía Confocal/métodos , Naftiridinas/metabolismo , Ploidias , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
10.
Chem Biol ; 19(8): 994-1000, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22921066

RESUMEN

To identify small molecules that selectively control hematopoietic stem cell differentiation, we performed an unbiased screen using primary human CD34(+) cells. We identified a plant-derived natural product, euphohelioscopin A, capable of selectively differentiating CD34(+) cells down the granulocyte/monocytic lineage. Euphohelioscopin A also inhibits proliferation and induces differentiation of the myeloid leukemia cell lines THP-1 and HL-60. Mechanistic studies revealed that euphohelioscopin A is an activator of protein kinase C (PKC), and that the promonocytic effects of this natural product are mediated by PKC activation. In addition to shedding insights into normal hematopoiesis, this work may ultimately facilitate the application of stem cell therapies to a host of myeloid dysfunctions.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diterpenos/farmacología , Proteína Quinasa C/metabolismo , Antígenos CD34/metabolismo , Linaje de la Célula , Células Cultivadas , Diterpenos/química , Granulocitos/citología , Células HEK293 , Células HL-60 , Células HeLa , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Indoles/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Maleimidas/farmacología , Proteína Quinasa C/química , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología
11.
Cell Stem Cell ; 10(2): 218-29, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22305571

RESUMEN

Clinical hematopoietic transplantation outcomes are strongly correlated with the numbers of cells infused. Anticipated novel therapeutic implementations of hematopoietic stem cells (HSCs) and their derivatives further increase interest in strategies to expand HSCs ex vivo. A fundamental limitation in all HSC-driven culture systems is the rapid generation of differentiating cells and their secreted inhibitory feedback signals. Herein we describe an integrated computational and experimental strategy that enables a tunable reduction in the global levels and impact of paracrine signaling factors in an automated closed-system process by employing a controlled fed-batch media dilution approach. Application of this system to human cord blood cells yielded a rapid (12-day) 11-fold increase of HSCs with self-renewing, multilineage repopulating ability. These results highlight the marked improvements that control of feedback signaling can offer primary stem cell culture and demonstrate a clinically relevant rapid and relatively low culture volume strategy for ex vivo HSC expansion.


Asunto(s)
Simulación por Computador , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Animales , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Medios de Cultivo/metabolismo , Retroalimentación Fisiológica , Sangre Fetal/citología , Humanos , Ratones , Ratones SCID , Comunicación Paracrina
12.
J Pharmacol Exp Ther ; 338(1): 318-27, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21493753

RESUMEN

The biological functions of the aryl hydrocarbon receptor (AHR) can be delineated into dioxin response element (DRE)-dependent or -independent activities. Ligands exhibiting either full or partial agonist activity, e.g., 2,3,7,8-tetrachlorodibenzo-p-dioxin and α-naphthoflavone, have been demonstrated to potentiate both DRE-dependent and -independent AHR function. In contrast, the recently identified selective AHR modulators (SAhRMs), e.g., 1-allyl-3-(3,4-dimethoxyphenyl)-7-(trifluoromethyl)-1H-indazole (SGA360), bias AHR toward DRE-independent functionality while displaying antagonism with regard to ligand-induced DRE-dependent transcription. Recent studies have expanded the physiological role of AHR to include modulation of hematopoietic progenitor expansion and immunoregulation. It remains to be established whether such physiological roles are mediated through DRE-dependent or -independent pathways. Here, we present evidence for a third class of AHR ligand, "pure" or complete antagonists with the capacity to suppress both DRE-dependent and -independent AHR functions, which may facilitate dissection of physiological AHR function with regard to DRE or non-DRE-mediated signaling. Competitive ligand binding assays together with in silico modeling identify N-(2-(1H-indol-3-yl)ethyl)-9-isopropyl-2-(5-methylpyridin-3-yl)-9H-purin-6-amine (GNF351) as a high-affinity AHR ligand. DRE-dependent reporter assays, in conjunction with quantitative polymerase chain reaction analysis of AHR targets, reveal GNF351 as a potent AHR antagonist that demonstrates efficacy in the nanomolar range. Furthermore, unlike many currently used AHR antagonists, e.g., α-naphthoflavone, GNF351 is devoid of partial agonist potential. It is noteworthy that in a model of AHR-mediated DRE-independent function, i.e., suppression of cytokine-induced acute-phase gene expression, GNF351 has the capacity to antagonize agonist and SAhRM-mediated suppression of SAA1. Such data indicate that GNF351 is a pure antagonist with the capacity to inhibit both DRE-dependent and -independent activity.


Asunto(s)
Compuestos Alílicos/farmacología , Indazoles/farmacología , Indoles/farmacología , Purinas/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/metabolismo , Compuestos Alílicos/metabolismo , Animales , Sitios de Unión/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Indazoles/metabolismo , Indoles/química , Indoles/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Purinas/química , Purinas/metabolismo
14.
Angew Chem Int Ed Engl ; 50(1): 200-42, 2011 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-21184400

RESUMEN

Potential applications of stem cells in medicine range from their inclusion in disease modeling and drug discovery to cell transplantation and regenerative therapies. However, before this promise can be realized several obstacles must be overcome, including the control of stem cell differentiation, allogeneic rejection and limited cell availability. This will require an improved understanding of the mechanisms that govern stem cell potential and the development of robust methods to efficiently control their fate. Recently, a number of small molecules have been identified that can be used both in vitro and in vivo as tools to expand stem cells, direct their differentiation, or reprogram somatic cells to a more naive state. These molecules have provided a wealth of insights into the signaling and epigenetic mechanisms that regulate stem cell biology, and are already beginning to contribute to the development of effective treatments for tissue repair and regeneration.


Asunto(s)
Medicina Regenerativa/métodos , Investigación con Células Madre , Células Madre/fisiología , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Humanos , Medicina Regenerativa/tendencias , Trasplante de Células Madre , Células Madre/citología , Ingeniería de Tejidos/tendencias
15.
Science ; 329(5997): 1345-8, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20688981

RESUMEN

Although practiced clinically for more than 40 years, the use of hematopoietic stem cell (HSC) transplants remains limited by the ability to expand these cells ex vivo. An unbiased screen with primary human HSCs identified a purine derivative, StemRegenin 1 (SR1), that promotes the ex vivo expansion of CD34+ cells. Culture of HSCs with SR1 led to a 50-fold increase in cells expressing CD34 and a 17-fold increase in cells that retain the ability to engraft immunodeficient mice. Mechanistic studies show that SR1 acts by antagonizing the aryl hydrocarbon receptor (AHR). The identification of SR1 and AHR modulation as a means to induce ex vivo HSC expansion should facilitate the clinical use of HSC therapy.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Purinas/metabolismo , Purinas/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Antígeno AC133 , Animales , Antígenos CD/análisis , Antígenos CD34/análisis , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Recuento de Células , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Citocromo P-450 CYP1B1 , Citocinas/farmacología , Glicoproteínas/análisis , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/fisiología , Péptidos/análisis , Dibenzodioxinas Policloradas/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas , Especificidad de la Especie
16.
Biochem Pharmacol ; 78(8): 966-73, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19481066

RESUMEN

Bz-423 is a pro-apoptotic 1,4-benzodiazepine with therapeutic properties in murine models of lupus demonstrating selectivity for autoreactive lymphocytes. Bz-423 modulates the F(1)F(0)-ATPase, inducing the formation of superoxide within the mitochondrial respiratory chain, which then functions as a second messenger initiating apoptosis. In order to understand some of the features that contribute to the increased sensitivity of lymphocytes, we report the signaling pathway engaged by Bz-423 in a Burkitt lymphoma cell line (Ramos). Following the generation of superoxide, Bz-423-induced apoptosis requires the activation of Bax and Bak to induce mitochondrial outer membrane permeabilization and cytochrome c release. Knockdown of the BH3-only proteins Bad, Bim, Bik, and Puma inhibits Bz-423 apoptosis, suggesting that these proteins serve as upstream sensors of the oxidant stress induced by Bz-423. Treatment with Bz-423 results in superoxide-dependent Mcl-1 degradation, implicating this protein as the link between Bz-423-induced superoxide and Bax and Bak activation. In contrast to fibroblasts, B cell death induced by Bz-423 is independent of c-Jun N-terminal kinase. These results demonstrate that superoxide generated from the mitochondrial respiratory chain as a consequence of a respiratory transition can signal a specific apoptotic response that differs across cell types.


Asunto(s)
Apoptosis/fisiología , Benzodiazepinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Avidina/metabolismo , Linfocitos B/metabolismo , Biotinilación , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colorantes/metabolismo , Electroporación , Fluoresceínas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Potenciales de la Membrana/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Propidio/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Superóxidos/metabolismo
17.
Free Radic Biol Med ; 45(9): 1232-42, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18718527

RESUMEN

Bz-423 is a proapoptotic 1,4-benzodiazepine with potent therapeutic properties in murine models of lupus and psoriasis. Bz-423 modulates the F(1)F(0)-ATPase, inducing the formation of superoxide within the mitochondrial respiratory chain, which then functions as a second messenger initiating apoptosis. Herein, we report the signaling pathway activated by Bz-423 in mouse embryonic fibroblasts containing knockouts of key apoptotic proteins. Bz-423-induced superoxide activates cytosolic ASK1 and its release from thioredoxin. A mitogen-activated protein kinase cascade follows, leading to the specific phosphorylation of JNK. JNK signals activation of Bax and Bak which then induces mitochondrial outer membrane permeabilization to cause the release of cytochrome c and a commitment to apoptosis. The response of these cells to Bz-423 is critically dependent on both superoxide and JNK activation as antioxidants and the JNK inhibitor SP600125 prevents Bax translocation, cytochrome c release, and cell death. These results demonstrate that superoxide generated from the mitochondrial respiratory chain as a consequence of a respiratory transition can signal a sequential and specific apoptotic response. Collectively, these data suggest that the selectivity of Bz-423 observed in vivo results from cell-type specific differences in redox balance and signaling by ASK1 and Bcl-2 proteins.


Asunto(s)
Apoptosis , Benzodiazepinas/farmacología , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , ADN/metabolismo , Diploidia , Potenciales de la Membrana , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Modelos Biológicos , Superóxidos
18.
J Pharmacol Exp Ther ; 313(1): 56-63, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15576471

RESUMEN

Bz-423 is a new benzodiazepine that has cytotoxic and cytostatic effects against a number of cell types in culture, and recent studies have shown efficacy in experimental lupus models in rodents. The present study demonstrates that treating human skin in organ culture with Bz-423 suppresses retinoid-induced epidermal hyperplasia. Bz-423 suppresses hyperplasia in organ culture at concentrations that also inhibit keratinocyte proliferation in monolayer culture but that are not cytotoxic for keratinocytes and do not inhibit fibroblast growth. Under conditions in which keratinocyte proliferation is inhibited, there is no measurable effect on epidermal growth factor receptor activation, but there is reduced signaling at the level of extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase. Suppression of keratinocyte growth by Bz-423 is associated with generation of intracellular oxidants. However, antioxidant treatment reduces keratinocyte cytotoxicity that occurs at high concentrations of Bz-423, but it does not inhibit growth suppression. Together, these data suggest that Bz-423 has the potential to limit the untoward effects associated with topical retinoid treatment, and in addition, may have therapeutic effects against other forms of epidermal hyperplasia.


Asunto(s)
Benzodiazepinas/farmacología , Queratinocitos/efectos de los fármacos , Retinoides/antagonistas & inhibidores , Retinoides/farmacología , Piel/patología , Comunicación Autocrina/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Receptores ErbB/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Humanos , Hiperplasia , Immunoblotting , Indicadores y Reactivos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Técnicas de Cultivo de Órganos , Comunicación Paracrina/efectos de los fármacos , Fosforilación , Especies Reactivas de Oxígeno , Transducción de Señal/efectos de los fármacos
19.
J Biol Chem ; 279(28): 29615-21, 2004 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-15123639

RESUMEN

Bz-423 is a 1,4-benzodiazepine with selective lymphotoxic properties and potent therapeutic activity against lupus-like disease in autoimmune mice. In NZB/W lupus-prone mice, Bz-423 specifically kills germinal center B cells, which are the cells that drive disease both in this model and in human systemic lupus erythematosus. In this report, the mechanistic basis for the selective action of Bz-423 is investigated. We show that Bz-423-induces superoxide as an immediate early response and that this reactive oxygen species is more effective as a second messenger death signal in B cells activated by B cell receptor stimulation compared with resting cells. As a result, low [Bz-423] that are not cytotoxic to non-stimulated cells kill stimulated cells in synergy with anti-immunoglobulin M antibodies. Subsequent experiments demonstrated that Bz-423 extends the rise in intracellular calcium that accompanies anti-immunoglobulin M stimulation, and this effect mediates the synergistic death response. Because B cell hyperactivation and altered calcium signaling is a distinguishing feature of autoreactive lymphocytes in lupus, the mechanism by which Bz-423 induces apoptosis preferentially targets disease-causing cells on the basis of their activation state. Thus, molecules like Bz-423 could form the basis for new and selective anti-lupus agents.


Asunto(s)
Apoptosis/fisiología , Linfocitos B/efectos de los fármacos , Benzodiazepinas/farmacología , Señalización del Calcio/fisiología , Ácido Egtácico/análogos & derivados , Activación de Linfocitos/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Benzodiazepinas/química , Benzodiazepinas/uso terapéutico , Calcio/metabolismo , Células Cultivadas , Quelantes/metabolismo , Modelos Animales de Enfermedad , Ácido Egtácico/metabolismo , Femenino , Colorantes Fluorescentes/metabolismo , Centro Germinal/citología , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Superóxidos/metabolismo
20.
Cancer Res ; 64(2): 696-703, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14744787

RESUMEN

Resveratrol (3,5,4-trihydroxystilbene), a natural phytoalexin present in grapes, nuts, and red wine, has antineoplastic activities. Several molecular mechanisms have been described to underlie its effects on cells in vitro and in vivo. In the present study, the response of ovarian cancer cells to resveratrol is explored. Resveratrol inhibited growth and induced death in a panel of five human ovarian carcinoma cell lines. The response was associated with mitochondrial release of cytochrome c, formation of the apoptosome complex, and caspase activation. Surprisingly, even with these molecular features of apoptosis, analysis of resveratrol-treated cells by light and electron microscopy revealed morphology and ultrastructural changes indicative of autophagocytic, rather than apoptotic, death. This suggests that resveratrol can induce cell death through two distinct pathways. Consistent with resveratrol's ability to kill cells via nonapoptotic processes, cells transfected to express high levels of the antiapoptotic proteins Bcl-x(L) and Bcl-2 are equally sensitive as control cells to resveratrol. Together, these findings show that resveratrol induces cell death in ovarian cancer cells through a mechanism distinct from apoptosis, therefore suggesting that it may provide leverage to treat ovarian cancer that is chemoresistant on the basis of ineffective apoptosis.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias Ováricas/patología , Estilbenos/toxicidad , Antineoplásicos Fitogénicos/toxicidad , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/toxicidad , Femenino , Humanos , Cinética , Microscopía Fluorescente , Plásmidos , Resveratrol
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...